13
Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation, and Inactivation Graphical Abstract Highlights d Notum is an extracellular Wnt deacylase d Acylation maintains Wnt folding and its active monomeric conformation d Notum in neuroectoderm is required for vertebrate neural and head induction d Distinct Wnt inactivation mechanisms by Notum and Tiki orchestrate brain development Authors Xinjun Zhang, Seong-Moon Cheong, ..., Jose Garcia Abreu, Xi He Correspondence [email protected] In Brief Wnt proteins require a lipid modification for receptor binding and activity. Zhang and Cheong et al. report that Notum, a secreted Wnt antagonist, is a Wnt deacylase that inactivates Wnt proteins and is required for vertebrate brain development. Deacylated Wnt forms oxidized-oligomers, suggesting that acylation is essential for Wnt structure. Accession Numbers KP781855 KP781856 KP781857 Zhang et al., 2015, Developmental Cell 32, 719–730 March 23, 2015 ª2015 Elsevier Inc. http://dx.doi.org/10.1016/j.devcel.2015.02.014

Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

Article

Notum Is Required for Neural and Head Induction via

Wnt Deacylation, Oxidation, and Inactivation

Graphical Abstract

Highlights

d Notum is an extracellular Wnt deacylase

d Acylation maintains Wnt folding and its active monomeric

conformation

d Notum in neuroectoderm is required for vertebrate neural and

head induction

d Distinct Wnt inactivation mechanisms by Notum and Tiki

orchestrate brain development

Zhang et al., 2015, Developmental Cell 32, 719–730March 23, 2015 ª2015 Elsevier Inc.http://dx.doi.org/10.1016/j.devcel.2015.02.014

Authors

XinjunZhang,Seong-MoonCheong, ...,

Jose Garcia Abreu, Xi He

[email protected]

In Brief

Wnt proteins require a lipid modification

for receptor binding and activity. Zhang

and Cheong et al. report that Notum, a

secreted Wnt antagonist, is a Wnt

deacylase that inactivates Wnt proteins

and is required for vertebrate brain

development. Deacylated Wnt forms

oxidized-oligomers, suggesting that

acylation is essential for Wnt structure.

Accession Numbers

KP781855

KP781856

KP781857

Page 2: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

Developmental Cell

Article

Notum Is Required for Neural and Head Inductionvia Wnt Deacylation, Oxidation, and InactivationXinjun Zhang,1,4 Seong-Moon Cheong,1,4 Nathalia G. Amado,1 Alice H. Reis,2 Bryan T. MacDonald,1 Matthias Zebisch,3

E. Yvonne Jones,3 Jose Garcia Abreu,2 and Xi He1,*1Department of Neurology, The F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston,

MA 02115, USA2Instituto de Ciencias Biomedicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil3Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK4Co-first author

*Correspondence: [email protected]://dx.doi.org/10.1016/j.devcel.2015.02.014

SUMMARY

Secreted Wnt morphogens are essential for em-bryogenesis and homeostasis and require a lipid/palmitoleoylate modification for receptor bindingand activity. Notum is a secreted Wnt antagonistthat belongs to the a/b hydrolase superfamily, butits mechanism of action and roles in vertebrateembryogenesis are not fully understood. Here, wereport that Notum hydrolyzes the Wnt palmitoleoy-late adduct extracellularly, resulting in inactivatedWnt proteins that form oxidized oligomers incapableof receptor binding. Thus, Notum is aWnt deacylase,and palmitoleoylation is obligatory for the Wnt struc-ture that maintains its active monomeric conforma-tion. Notum is expressed in naive ectoderm and neu-ral plate in Xenopus and is required for neural andhead induction. These findings suggest that Notumis a prerequisite for the ‘‘default’’ neural fate andthat distinct mechanisms of Wnt inactivation by theTiki protease in the Organizer and the Notum de-acylase in presumptive neuroectoderm orchestratevertebrate brain development.

INTRODUCTION

TheWnt family of secreted lipoproteins controls animal develop-

ment including axial patterning and cell fate specification and

governs tissue homeostasis and stem cell renewal (Clevers

and Nusse, 2012; MacDonald et al., 2009). Anomalies in Wnt

signaling cause human diseases including birth defects, cancer,

and osteoporosis (Clevers and Nusse, 2012; MacDonald et al.,

2009). Wnt proteins engage multiple transmembrane receptors,

including the Frizzled (Fz) serpentine receptors and low-density

lipoprotein receptor-related proteins 5 and 6 (LRP5/6), which

induce stabilization of the transcription co-activator b-catenin

(MacDonald and He, 2012). Wnt proteins act locally near the

source of their secretion in many contexts, and they also behave

as morphogens with long range signaling properties (Hausmann

et al., 2007; Strigini and Cohen, 2000; Zecca et al., 1996; but see

Develo

Alexandre et al., 2014). Critical for these versatile signaling prop-

erties is a lipid modification of Wnt proteins referred to as O-pal-

mitoleoylation (Takada et al., 2006; Willert et al., 2003). This form

of O-acylation, which has been best demonstrated for themouse

Wnt3a, conjugates a mono-unsaturated palmitoleic acid onto

the hydroxyl group of a conserved serine residue (serine 209 of

Wnt3a), likely through the action of a Wnt-specific O-acyltrans-

ferase called Porcupine in the ER (Rios-Esteves et al., 2014; Ta-

kada et al., 2006). Wnt palmitoleoylation serves two essential

functions. First, palmitoleoylation appears to be obligatory for

Wnt secretion, as the Wnt3a(S209A) mutant, which has serine

209 substituted by an alanine, and thus lacks palmitoleoylation,

is not secreted (Takada et al., 2006) possibly as a result of failure

to bind to Wntless, a Wnt chaperone in the secretory pathway

(Coombs et al., 2010; Herr and Basler, 2012; Tang et al., 2012).

Second, the lipid modification is required for secreted Wnt li-

gands to signal, as the palmitoleoylate adduct inserts into a hy-

drophobic cleft of the Fz receptor to form one of the two Wnt-Fz

binding interfaces (Janda et al., 2012).

Canonical Wnt signaling plays multiple roles including axial

patterning and germ layer specification in vertebrate embryo-

genesis (De Robertis and Kuroda, 2004; Hikasa and Sokol,

2013; Stern, 2005). In Xenopus embryos, maternal Wnt/b-cate-

nin signaling promotes the dorsal Spemann-Mangold Organizer

and dorso-ventral (DV) axis formation (Harland and Gerhart,

1997). During gastrulation, a gradient of Wnt/b-catenin signaling

occurs along the anterio-posterior (AP) axis, with higher levels

posteriorly (Kiecker and Niehrs, 2001). The Organizer pro-

motes head development via secreting Wnt antagonists such

as secreted frizzled-related proteins (sFRPs) and Dickkopf-1

(Dkk1), which bind to and inhibit Wnt/Fz and LRP6, respectively

(Cruciat and Niehrs, 2013; De Robertis and Kuroda, 2004). We

recently identified another Organizer-specific and membrane-

tethered Wnt antagonist, Tiki, which is a prototypic Wnt inacti-

vating protease and is required for head formation (Zhang

et al., 2012). The Organizer is also essential for neural induction.

This has been primarily attributed to Organizer-secreted bone

morphogenetic protein (BMP) antagonists such as Chordin and

Noggin, which shield the naive ectoderm from the influence of

BMPs that promote epidermal differentiation, thereby permitting

‘‘default’’ neuralization (De Robertis and Kuroda, 2004; Ozair

et al., 2013; Stern, 2005). Evidence suggests that inhibition of

Wnt signaling and active fibroblast growth factor (FGF) signaling

pmental Cell 32, 719–730, March 23, 2015 ª2015 Elsevier Inc. 719

Page 3: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

are also required for neural induction in Xenopus and chick em-

bryos (Delaune et al., 2005; Fuentealba et al., 2007; Heeg-Trues-

dell and LaBonne, 2006; Kengaku and Okamoto, 1995; Lamb

and Harland, 1995; Marchal et al., 2009; Pera et al., 2003; Stern,

2005; Wilson et al., 2001). But how regulation of Wnt signaling is

achieved and contributes to neural induction by theOrganizer re-

mains unknown.

Notum (or Wingful) is a secreted antagonist of Wingless (Wg,

Drosophila Wnt1) (Gerlitz and Basler, 2002; Giraldez et al.,

2002). Sequence analysis places Notum in the so-called a/b hy-

drolase superfamily that includes various hydrolytic enzymes

(Nardini and Dijkstra, 1999). Notum appears to regulate Wg

extracellular distribution during wing development (Gerlitz and

Basler, 2002; Giraldez et al., 2002). As heparan sulfate proteogly-

cans, in particular glypicans Dally and Dally-like protein (Dlp), are

involved inmodulating theWg gradient (Han et al., 2005; Yan and

Lin, 2009), and as Notum is in sequence most similar to plant

pectin acetylesterases, Notum was first proposed to modify

glycosaminoglycans (long unbranched polysaccharides) in Dally

and Dlp (Giraldez et al., 2002). Subsequent experiments led to a

revised model that Notum cleaves the glycosylphosphatidylino-

sitol (GPI) anchor of Dlp, thereby switching Dlp from a mem-

brane-anchored activator into a secreted antagonist (Kreuger

et al., 2004). But because Dlp and Dally participate in functions

of most or all morphogen families and play both positive and

negative roles (Beckett et al., 2008; Filmus et al., 2008; Yan

and Lin, 2009), the model that Notum antagonizes Wg signaling

via modifying Dlp is controversial. Notum is conserved from in-

vertebrates to human (Giraldez et al., 2002). In planarians, Notum

and Wnt govern head and tail regeneration, respectively (Pe-

tersen and Reddien, 2011), reflecting a common theme of Wnt

antagonists versus Wnt in AP patterning. In zebrafish, Notum

has a role in DV patterning of the neural tube (Flowers et al.,

2012). But the roles of Notum in early vertebrate embryogenesis

are unknown. Here, we report that Notum is a Wnt-inactivating

deacylase and is critical for neural and head induction by acting

within the presumptive neuroectoderm.

RESULTS

Notum Is a Specific Wnt Antagonist in VertebrateEmbryos and Mammalian CellsThe mouse or human genome harbors a single Notum gene.

Expression of the mouse Notum in HEK293T cells produced

secreted Notum in the conditioned medium (CM) (see below),

and inhibited a prototypic Wnt-responsive TOPFLASH reporter

induced by either co-expression of mouse Wnt3a or treatment

with recombinant human WNT3A proteins (Figures 1A and 1B).

Thus Notum inhibits Wnt3a extracellularly. Notum shares with

other a/b hydrolase proteins a conserved motif GxSxG, in which

the serine mediates the hydrolytic reaction (Nardini and Dijkstra,

1999). Notum(S239A), in which the serine was replaced by an

alanine, was unable to inhibit Wnt3a signaling (Figure 1C),

consistent with results that an analogous Drosophila Notum

mutant fails to inhibit Wg (Giraldez et al., 2002). Thus the enzy-

matic activity of Notum appears to be obligatory for Wg/Wnt

antagonism.

Notum inhibited Wnt signaling in Xenopus laevis embryos.

Ventral injection of Xenopus Wnt8 or b-catenin mRNA into

720 Developmental Cell 32, 719–730, March 23, 2015 ª2015 Elsevier

4-cell embryos resulted in axis duplication, and co-injection of

mouse Notum mRNA, but not Notum(S239A) mRNA, inhibited

axis duplication by Wnt8 (Figure 1D). Notum did not inhibit axis

duplication by b-catenin (Figure 1D), consistent with Notum

acting extracellularly. In animal cap explants, Notum inhibited

Wnt8, but not b-catenin, induced expression of Xnr3, a b-catenin

target gene and dorsal marker (Figures 1E and 1F). Importantly,

Notum showed no effect on induction of Xbra, a mesodermal

marker, by Nodal/Xnr1 (a transforming growth factor-bmember)

or basic FGF (bFGF), or induction of Vent2, a ventral marker, by

BMP4 (Figures 1G–1I). These data demonstrate that Notum is a

specific Wnt antagonist, consistent with phenotypic analyses in

invertebrates (Gerlitz and Basler, 2002; Giraldez et al., 2002; Pe-

tersen and Reddien, 2011) and zebrafish (Flowers et al., 2012).

Notum Modifies and Inactivates Wnt ProteinsNotum has been suggested to cleave the GPI anchor of Dlp in

flies (Kreuger et al., 2004). Of the six glypicans encoded in the

mouse/human genome, Glypican 3 (GPC3) and GPC4 have

been implicated in Wnt pathways (Capurro et al., 2014; Filmus

et al., 2008; Sakane et al., 2012). However, in HEK293T cells,

the mouse Notum exhibited minimal GPI-cleaving activity on

GPC3 or GPC4 when compared to a positive control, phospha-

tidylinositol-specific phospholipase C (PI-PLC), which cleaved

the GPI of both glypicans, shedding them into CM (Figures

S1A and S1B).

We previously identified Tiki as a prototypic Wnt modifying

and inactivating enzyme (Zhang et al., 2012). While the Wnt3a

protein is hydrophobic because of its lipid modification (Willert

et al., 2003), Wnt3a modified by Tiki is hydrophilic, as we

demonstrated in a Triton X-114 detergent-aqueous phase sep-

aration assay (Zhang et al., 2012). This observation led to the

suspicion that Tiki might have Wnt deacylase activity, but this

was ruled out by metabolic labeling showing that Wnt3a palmi-

toleoylation was unaltered by Tiki (Zhang et al., 2012). Notum

shares the distinction with Tiki as the only established Wnt

antagonists with enzymatic motifs, raising the possibility that

Notum may modify Wnt proteins. Indeed, Wnt3a when co-ex-

pressed with Notum, like when co-expressed with Tiki, became

predominantly hydrophilic as demonstrated by the Triton X-114

phase separation assay (Figure 2A). Correlating with the in-

ability to inhibit Wnt3a signaling (Figure 1C), Notum(S239A)

did not alter Wnt3a hydrophobicity (Figure 2A). None of several

other secreted or secretory pathway enzymes that exhibit de-

palmitoylation activities including palmitoyl protein thioes-

terases (Zeidman et al., 2009), or are genetically implicated in

Wnt-regulated events such as secreted phospholipase A2

(Cormier et al., 1997), was able to alter Wnt3a hydrophobicity

(data not shown). Therefore, the ability of Notum to modify

Wnt3a hydrophobicity, like that of Tiki, is specific and depen-

dent on its hydrolase motif.

Wnt-induced phosphorylation of the LRP6 co-receptor and

Dishevelled (Dvl), a component downstreamof Fz, indicates acti-

vation of transmembrane signaling (MacDonald and He, 2012).

Wnt3a CM from control HEK293T cells or cells expressing No-

tum(S239A) induced phosphorylation of LRP6, Dvl2, and the

TOPFLASH reporter, but Wnt3a CM from cells that co-ex-

pressed Notum failed to do so (Figures 2B and 2C), implying

Wnt3a inactivation by Notum. But under this condition, secreted

Inc.

Page 4: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

Figure 1. NotumAntagonizesWnt Signaling

in Mammalian Cells and Xenopus Embryos

(A) Notum inhibited TOPFLASH induced byWnt3a

in HEK293T cells. Increasing doses of a Notum

expression vector were co-transfected with an

expression vector for Wnt3a.

(B) Notum inhibited TOPFLASH induced by the

recombinant WNT3A protein. HEK293T cells

transfected with increasing doses of the Notum

expression vector were incubated with the

WNT3A protein.

(C) Notum(S239A) lacked the ability to inhibit

TOPFLASH induced by the WNT3A protein.

(D) Notum inhibited axis duplication induced by

Xenopus Wnt8, but not b-catenin, while No-

tum(S239A) did not inhibit axis duplication

induced by either Wnt8 or b-catenin. n, embryos

examined.

(E–I) Notum in animal pole explants inhibited

expression of Xnr3 induced byWnt8 (E), but not by

b-catenin (F), nor did it inhibit Xbra expression

induced by Nodal/Xnr1 (G), or bFGF (H), or Vent2

expression induced by BMP4 (I). EF1a, a loading

control; uninjected embryos, uninj.; whole em-

bryos, WE; whole embryos without the reverse

transcriptase, -RT. There were two doses of

Notum mRNA that were injected.

Error bars (A–C) represent SD of triplicated ex-

periments.

See also Figure S1.

Notum and Wnt3a proteins co-existed in the CM (Figure 2B).

To this end, we generated Notum-TM, which fuses Notum to

a transmembrane domain at the carboxyl terminus and is

anchored on the plasma membrane. Notum-TM was undetect-

able in CM (Figure 2B), but diminished Wnt3a hydrophobicity

and inhibited Wnt3a-induced TOPFLASH as Notum did (Figures

2A and S2A). Wnt3a CM from cells co-expressing Notum-TM, in

contrast to Wnt3a CM from cells co-expressing Notum(S239A)-

TM, induced neither phosphorylation events nor TOPFLASH

(Figures 2B and 2C), suggesting thatWnt3a had been inactivated

by Notum or Notum-TM. Wnt3a from Notum-expressing cells

exhibited poor binding to Fz, as examined using pull-downs

by Fz8 extracellular cysteine-rich domain fused with IgG-Fc

(Fz8CRD-Fc), whereas Wnt3a from Notum(S239A) cells showed

normal Fz binding (Figure 2D). Therefore, Wnt3a modified by

Developmental Cell 32, 719–73

Notum, like that modified by Tiki (Zhang

et al., 2012), is not competent to bind to

the Wnt receptor.

Notum Causes WntDepalmitoleoylationNotum, like Tiki, had no appreciable ef-

fect on Wnt3a secretion, but resulted in

the Wnt3a protein exhibiting faster

mobility during gel electrophoresis (Fig-

ure 2B) (Zhang et al., 2012), consistent

with Wnt3a being modified by Notum.

Tiki cleavage of eight residues at Wnt3a

amino terminus causes the change in

Wnt3a hydrophobicity and electropho-

retic mobility (Zhang et al., 2012). However, unlike Tiki, Notum

did not exhibit amino terminal cleavage activity toward Wnt3a

(Figure S2B). Thus, despite the fact that Wnt3a modified by

Notum and Tiki behaved similarly, Notum is unlikely to be a

Wnt protease, and therefore modifies Wnt3a in a fundamentally

different manner.

Mass spectrometry of Wnt3a and the Wnt8 crystal structure

suggest that O-palmitoleoylation and N-glycosylation are the

predominant modifications of an active Wnt protein (Janda

et al., 2012; Takada et al., 2006; Zhang et al., 2012). Others

and we have shown that glycosylation/de-glycosylation does

not alter Wnt3a hydrophobicity (Komekado et al., 2007; Zhang

et al., 2012). These considerations raise the possibility that

Notum may be a Wnt deacylase that removes palmitoleic acid

through hydrolysis, thereby inactivating Wnt. We performed

0, March 23, 2015 ª2015 Elsevier Inc. 721

Page 5: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

Figure 2. Notum Causes Wnt Deacylation

and Inactivation

(A) The Wnt3a protein modified by Notum became

hydrophilic in the Triton X-114 detergent-aqueous

phase separation assay. Wnt3a from mock or

Notum(S239A)-expressing cells was hydrophobic

and partitioned in the detergent (De) phase, but

Wnt3a from Notum- or Notum-TM-expressing

cells partitioned in the aqueous (Aq) phase; total

input, T.

(B) Wnt3a CM from Notum- or Notum-TM-ex-

pressing cells was inactive and induced minimal

phosphorylation of LRP6 and Dvl2 in mouse

embryonic fibroblast cells, whereas Wnt3a CM

from mock, Notum(S239A)-, or Notum(S239A)-

TM-expressing cells induced phosphorylation

of LRP6 and Dvl2. Note that Wnt3a from Notum-

or Notum-TM-expressing cells exhibited slightly

faster migration. WCL, whole cell lysates.

(C) Wnt3a CM from Notum or Notum-TM-ex-

pressing cells induced minimal TOPFLASH in

HEK293T cells, whereas Wnt3a CM from mock,

Notum(S239A)-, or Notum(S239A)-TM-expressing

cells induced TOPFLASH. Error bars represent SD

of triplicated experiments.

(D) Wnt3a secreted from Notum-expressing cells

exhibited minimal binding to mFz8CRD-IgG,

whereas Wnt3a secreted from mock or No-

tum(S239A)-expressing cells exhibited binding.

(E) Notum, but not Notum(S239A), reduced Wnt3a

acylation when they were co-expressed in

HEK293T cells.

See also Figure S2.

metabolic labeling of Wnt3a with a palmitic acid analog (az-15)

(Zhang et al., 2012), which can be desaturated and incorporated

into Wnt3a in the cell (Hannoush, 2012; Rios-Esteves and Resh,

2013; Zhang et al., 2012). The resulting lipidation of Wnt3a can

be detected using a fluorescent dye through click chemistry

(Zhang et al., 2012). Indeed, Wnt3a CM from control or No-

tum(S239A)-expressing cells exhibited strong lipid labeling indi-

cating palmitoleoylation, but Wnt3a CM from Notum-expressing

cells lacked lipid labeling (Figure 2E). Therefore, Notum through

its hydrolase motif causes Wnt3a depalmitoleoylation.

Notum Is Likely a Wnt Deacylase and ActsExtracellularlyWnt palmitoleoylation appears to be essential for Wnt secretion,

as Wnt3a(S209A), which lacks the palmitoleoylated serine, is not

secreted, nor are any of the Wnt proteins that are expressed in

cells/embryos deficient for Porcupine (Barrott et al., 2011; Bie-

722 Developmental Cell 32, 719–730, March 23, 2015 ª2015 Elsevier Inc.

chele et al., 2011; Proffitt and Virshup,

2012; Takada et al., 2006). Notum does

not affect Wnt3a secretion (Figure 2B),

suggesting that depalmitoleoylation oc-

curs after Wnt3a secretion. Indeed, in

contrast to secreted Wnt3a (Figure 2A),

hydrophobicity of Wnt3a from cell lysates

was less affected by Notum (Figure 3A),

suggesting that Wnt3a in the secretory

pathway is insensitive to Notum. Impor-

tantly, incubation of recombinantWNT3A in NotumCM in culture

resulted in loss ofWnt3a hydrophobicity (Figure 3B). This result is

consistent with Notum inhibition of recombinant WNT3A pro-

teins added to responding cells (Figure 1B). We purified, from

respective CM, secreted Notum, Notum(S239A), and Wnt3a

that had been metabolically labeled by the palmitic acid analog

and reconstituted the depalmitoleoylation reaction in vitro.

Notum, but not Notum(S239A), caused Wnt3a deacylation (Fig-

ure 3C). The simplest interpretation of these results is that Notum

is a Wnt deacylase.

Palmitoleoylation Is Required for the Wnt Structure andActive Monomeric StateWnt3a cleaved by Tiki retains normal palmitoleoylation, but is hy-

drophilic (Zhang et al., 2012). This paradox appears to be ex-

plained by our finding that Tiki-cleaved Wnt3a (or an engineered

Wnt3aDN that lacks the eight amino terminal residues removed

Page 6: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

Figure 3. Notum Is Likely a Wnt Deacylase Acting Extracellularly and Causes Wnt3a and Wnt5a to Form Oxidized Oligomers

(A) Notum had minimal effects on hydrophobicity of Wnt3a from WCL in the detergent-aqueous phase separation assay.

(B) Recombinant WNT3A proteins lost hydrophobicity after incubation with Notum-expressing cells.

(C) Notum reduced Wnt3a acylation in vitro. Purified metabolically labeled Wnt3a protein was incubated with mock, purified Notum, or Notum(S239A)

proteins.

(D) Wnt3a secreted from Notum-, but not Notum(S239A)-, expressing cells formed oxidized oligomer. Wnt3a CM from mock, Notum-, or Notum(S239A)-ex-

pressing cells was analyzed by non-reducing or reducing SDS-PAGE. Wnt3a monomers and oxidized oligomers were labeled by an arrow and asterisk,

respectively.

(E) Recombinant WNT3A proteins formed oxidized oligomers upon incubation with Notum-, but not mock or Notum(S239A)-,expressing cells.

(F) Wnt5a proteins secreted from Notum-expressing cells formed oxidized oligomers.

See also Figure S2.

by Tiki) forms large, but soluble, Wnt3a oligomers that are linked

by inter-Wnt3a disulfide bonds (Zhang et al., 2012). These

oxidizedWnt3a oligomers behave in a strictly hydrophilicmanner

in the Triton X-114 phase separation assay, and thus may have

buried the lipid adduct inside the oligomer (Zhang et al., 2012).

It is worth noting that Wnt3a amino terminal residues cleaved

by Tiki do not contain a cysteine, yet their absence profoundly

alters Wnt3a structural integrity and disulfide bond patterns

(Zhang et al., 2012). Unexpectedly, Wnt3a modified by Notum

behaved in a similar manner, it formed soluble and large Wnt3a

oligomers that were linked by inter-Wnt3a disulfide bonds and

detected under non-reducing conditions (Figure 3D). Wnt3a

CM from control or Notum(S239A)-expressing cells yielded

mostly monomeric proteins under the same non-reducing condi-

tion (Figure 3D). This conversion of the active/monomeric form

Develo

into inactive/oxidized oligomeric formsbyNotumoccurred extra-

cellularly, because recombinant WNT3A, which is active/mono-

meric, became oxidized oligomers upon incubation in vitro with

Notum, but not with Notum(S239A), CM (Figure 3E). Such a con-

version after depalmitoleoylation by Notum was not a unique

property of Wnt3a, Wnt5a appeared to be deacylated and lose

its hydrophobicity upon co-expression with Notum (Figure S2C)

and was converted concomitantly from monomers to oxidized

oligomers (Figure 3F). Therefore, palmitoleoylation is essential

for the Wnt structure that ensures its active monomeric state.

Notum Is Expressed Dynamically during Xenopus

EmbryogenesisNotum functions in vertebrates have been studied in zebrafish,

which have three Notum genes, Notum1a, Notum1b, and

pmental Cell 32, 719–730, March 23, 2015 ª2015 Elsevier Inc. 723

Page 7: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

Figure 4. NotumExpression Patterns during

Xenopus Embryogenesis

(A and B) RT-PCR revealed that Notum is mater-

nally and zygotically expressed throughout Xen-

opus embryogenesis (A), and is enriched animally

and dorsally at stage 10.5; animal caps, AC; dorsal

marginal zone, DMZ; ventral marginal zone, VMZ;

vegetal caps, VC; a pan-mesodermal marker,

Xbra; a dorsal marker, Chordin; an animal and

ventral marker, Msx1.

(C–T) Whole mount in situ hybridization for Notum/

Notum’ expression, showing lateral view at 4-cell

and stage 6.5 (C and D); lateral and bisected view

at stage 8.5 (E and F); bisected view at stage 9.5

(G); lateral and bisected view at stage 10.5, with

dorsal on right (H and I); dorsal and bisected view

at the stage 11 (J and K); with an enlarged anterior

view showing stronger expression anteriorly (L);

dorso-anterior (M); dorsal (N, anterior on top); and

cross-section (O, dorsal on top; and P, an enlarged

view) at stage 15; lateral view at the tail bud stage

(Q); with an enlarged anterior view indicating

expression in the cement gland (R, arrow); lateral

view at the early tadpole stage (S); and with an

enlarged view indicating expression in the pro-

nephros region (T, arrowhead). The blue color at

the blastocoel surface in bisections was non-

specific.

See also Figure S3.

Notum2 (Cantu et al., 2013; Flowers et al., 2012). Notum1a and

1b are paralogs due to teleost genome duplication and are

orthologs of the mammalian/Drosophila Notum, and indeed,

Notum1a antagonizes Wnt/b-catenin signaling (Flowers et al.,

2012). Depletion of Notum1a causes neural tube defects in DV

patterning (Flowers et al., 2012). Notum2 is a distant relative of

mammalian/Drosophila Notum and does not appear to have

Wnt antagonist activity (Cantu et al., 2013).Notum2 is strictly ex-

pressed in muscle pioneer cells in larvae and has a role in axon

guidance during primary motor neurogenesis (Cantu et al.,

2013), which is unique to teleosts and amphibians (to endow

larvae with escape responses from predators, for example).

The Xenopus laevis (allotetraploid) genome contains Notum

and Notum’, which are pseudo-alleles due to genome duplica-

tion, and Notum2 (Figure S3). Notum and Notum’ are 94% iden-

tical in nucleotide (and corresponding amino acid) sequences

and are orthologs of the mammalian/Drosophila Notum and

fish Notum1a, whereas Notum2 is the ortholog of fish Notum2

(Figure S3). The Xenopus tropicalis (diploid) genome contains a

single Notum gene, and Notum2 (Figure S3).

Notum and Notum’ mRNAs are expressed in the egg (and

are enriched in the animal half, data not shown) and through

cleavage to gastrulation stages (Figure 4A) and are enriched

in the animal (prospective ectoderm) and dorsal regions

in early gastrula (Figure 4B). Whole mount in situ hybridiza-

tion showed expression of Notum and Notum’ (as the two

pseudo-alleles were likely cross-hybridized by the probe) in an-

724 Developmental Cell 32, 719–730, March 23, 2015 ª2015 Elsevier Inc.

imal blastomeres at 4-cell and stage 6.5

(Figures 4C and 4D). At stages 8.5 and

9.5 (blastula), Notum mRNA was de-

tected broadly in the animal region

(Figures 4E–4G). At stage 10 (early gastrula), Notum mRNA re-

mains broadly expressed animally, but was also detected in

the dorsal marginal zone (the Organizer), with lower expression

in the ventral marginal zone (Figures 4H and 4I). At stage

11, Notum mRNA was found in the forming neural plate in a

noticeable AP gradient (anterior high and posterior low), with

additional weaker expression in the head mesoderm (Figures

4J–4L). Notum mRNA remains detectable, but becomes faint

in the neural plate at stage 13 (data not shown). By stage 15,

Notum mRNA was detected at the anterior border of the neural

plate and in ventro-lateral epidermis excluding the neural plate

(Figures 4M and 4N). Cross-section of a stage 15 embryo

showed Notum expression in the lateral surface of the

epidermis and the lateral plate mesoderm (Figures 4O and

4P). Later, Notum was detected in the cement gland (an ante-

rior organ) at tail bud stages (stage 25, Figures 4Q and 4R), in

branchial arches, the otic vesicle, and developing pronephros,

with diffused expression in the head (stage 35, Figures 4S and

4T). Thus, Notum mRNA exhibits dynamic expression during

Xenopus embryogenesis, in particular during neural induction

and AP patterning.

Notum Is Required for Head FormationXenopus Notum and Notum’ behaved identically as the mouse

Notum in Wnt deacylation and inactivation (Figure S4). Dorsal

injection of synthetic mRNA for the mouse Notum, but not

Notum(S239A), induced an enlarged head as seen for Tiki1 or

Page 8: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

Figure 5. Notum Is Required for Anterior Development in Xenopus Embryos(A and B) Dorsal injection of Notum mRNA, but not Notum(S239A), induced an enlarged head similar to that induced by Tiki1 mRNA, and resulting phenotypes

were tabulated.

(C) The NotumMO and the Notum’MO inhibited protein synthesis from Xenopus Notum (xNotum) or Notum’ (xNotum’), but not mouse Notum (mNotum) mRNA.

Co, MO, and MO’ indicate control MO and MOs against xNotum and xNotum’, respectively.

(D and E) Dorso-animal injection of the two NotumMOs together caused anterior defects that were rescued bymNotum mRNA, and resulting phenotypes were

tabulated.

(F) The Notum MOs suppressed expression of forebrain markers Otx2 and Bf1 and a hindbrain marker Krox20 at stage 17. See also Table S1.

(G) Illustration of the MO-injected A1 and B1 blastomeres at 32-cell stage and their descendent tissues at stage 10.5.

(H) The Notum MOs injected into the A1 blastomeres caused anterior defects in more embryos (69%) than those injected into the B1 blastomeres (22%).

See also Figures S4 and S5; Tables S2 and S3.

Dkk1 mRNA injection (Figures 5A and 5B) (Zhang et al., 2012),

consistent withWnt inhibition in embryos.We designed twomor-

pholino antisense oligonucleotides (MOs) against the 50 regionsurrounding the ATG initiation codon of Notum and Notum’.

These two MOs blocked protein synthesis from mRNAs for Xen-

opus Notum andNotum’, respectively, but not themouseNotum

(Figure 5C) that does not have the MO-targeting sequence,

demonstrating the specificity of these MOs. Dorso-animal injec-

tion of the two MOs together, but not of a control MO, at the

8-cell stage caused severe deficiency in head formation, result-

ing in embryos lacking the forebrain, eyes, and the cement gland

(Figures 5D and 5E). These anterior defects were rescued when

theMOs were co-injected with mouseNotummRNA (Figures 5C

Develo

and 5E), illustrating that the phenotypes were a result of lacking

Notum. These anterior defects by the Notum MOs were also

rescued/over-rescued by co-injection of Dkk1 mRNA (Fig-

ure S5A) and by co-injection of a b-catenin MO that depletes

the b-catenin protein (Heasman et al., 2000) (Figures S5B and

S5C), supporting the notion that the defects were due to exces-

sive Wnt/b-catenin signaling. The overt anterior deficiencies by

the Notum MOs were accompanied by, at stage 17, diminished

expression of anterior markers Otx2, Bf1, Pax6 (forebrain), and

XAG (the cement gland), and of a hindbrain marker Krox20,

each of which was rescued by co-injection of the mouse Notum,

but notNotum(S239A), mRNA (Figures 5F, S5D, and S5E; Tables

S1 and S2). The expression of the spinal cord markerHoxb9was

pmental Cell 32, 719–730, March 23, 2015 ª2015 Elsevier Inc. 725

Page 9: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

unaffected by the Notum MOs (Figure S5E). Thus, like Tiki1,

Notum is required for Xenopus head patterning.

Notum Functions in Prospective EctodermTiki1 is specifically expressed in the Organizer, in particular, the

so-called head organizer that is composed of endomesodermal

tissues underlying the future forebrain (Zhang et al., 2012). In

Tiki1-depleted embryos, the head organizer function is signifi-

cantly compromised, as evidenced by diminished expression

of all head organizer markers examined, including Dkk1, Chor-

din, Lim1, Goosecoid, and Otx2, while the expression of other

dorsal markers such as Xnr3 and Xnot1 is unaffected (Zhang

et al., 2012). However, in Notum-depleted embryos, the expres-

sion of head organizer markers Chordin andGoosecoid, like that

of Xnr3 and Xnot1, was unchanged (Figure S5F; Table S3). Thus,

unlike Tiki1, Notum function appears to minimally impact the

head organizer integrity and may therefore participate in head

patterning by acting within the prospective ectoderm, in which

Notum is expressed during blastula and gastrula stages (Figures

4E–4L).

To examine this issue further, we injected, at the 32-cell stage,

Notum MOs into the dorsal A1 blastomeres, which give rise

mostly to the future head and neuro-ectoderm, or the dorsal

B1 blastomeres, which are fated primarily to become the dorsal

Organizer (Dale and Slack, 1987; Moody, 1987) (Figure 5G).

Notum depletion in A1 and B1 progenies showed drastically

different outcomes and resulted in head deficiencies in 69%

and 22% of embryos, respectively (Figure 5H), supporting the

notion that Notum is required primarily in the prospective ecto-

derm for anterior neural development.

Notum Is Required for Neural InductionInhibition of Wnt signaling has been suggested to be critical for

neural induction in Xenopus (Fuentealba et al., 2007; Heeg-

Truesdell and LaBonne, 2006), but the profound effect of Wnt

signaling on dorsal Organizer, which induces neural tissues

through secreting BMP antagonists, complicates studies on

this issue (Stern, 2005). Interestingly, embryos injected with

the Notum MOs, but not the control MO, showed a significant

reduction of a pan-neural marker Sox2, but an expansion of

the epidermal marker cytokeratin, and these reciprocal changes

were rescued by co-injection of the mouse Notum, but not

Notum(S239A), mRNA (Figure 6A; Table S4). Because Organizer

markers were unaffected (Figure S5E), these results suggest

that Notum acts within prospective ectoderm for neural

specification.

To examine this issue directly, we employed a classical neural

induction assay using animal cap explants. Chordin or Noggin

induced pan and anterior neural markers and suppressed

the epidermal marker in explants from control embryos, but

failed to do so in explants from Notum-depleted embryos, and

this neural induction defect was rescued by the mouse Notum,

but notNotum(S239A), mRNA (Figures 6B andS6A). Importantly,

the ability of Chordin to induce neural tissue was restored when

the b-catenin MO was co-injected with the Notum MOs (Fig-

ure 6C), suggesting the failure of neural induction upon Notum

depletion as a result of excessive Wnt/b-catenin signaling.

To rule out the possibility that Notum is required for the BMP

antagonists to function properly, we showed that Chordin or

726 Developmental Cell 32, 719–730, March 23, 2015 ª2015 Elsevier

Noggin inhibited BMP4 induction of the ventral Vent2 regardless

of Notum depletion (Figure S6B). Thus, Notum suppression of

Wnt signaling is a prerequisite for neural induction by BMP an-

tagonists. Indeed, overexpression of Notum, like that of Dkk1

(Fuentealba et al., 2007; Glinka et al., 1998), induced pan and

anterior neural markers in animal explants (Figure 6D).

DISCUSSION

Notum as a Wnt Deacylase: Comparisons with the WntProtease TikiGenetic analyses in planarian and zebrafish show that Notum in-

hibits signaling byWnt, but not byHedgehog or other growth fac-

tors (Flowers et al., 2012; Petersen andReddien, 2011).We show

that the mouse or Xenopus Notum is a Wnt3a antagonist in

mammalian cells and inhibits target gene activation by Wnt8,

butnotBMP,Nodal, or FGF inXenopusanimal explants (Figure1).

Notum belongs to the a/b hydrolase superfamily that includes

peptidases, lipases, esterases, and other hydrolytic enzymes

(Nardini and Dijkstra, 1999). It was proposed that Notum inhibits

Wg signaling via hydrolyzing the GPI anchor of the Dlp glypican

(Kreuger et al., 2004). But this model is strongly challenged by

the fact that Dlp, Dally, and their homologs participate in most

or all morphogen signaling pathways (Filmus et al., 2008).

Compared to PI-PLC, we detected minimal Notum cleavage of

the GPI anchor of GPC3 or GPC4 (Figure S1), two mammalian

glypicans that have been implicated in Wnt pathways (Capurro

et al., 2014; Filmus et al., 2008; Sakane et al., 2012).

Tiki is an archetypal Wnt antagonist that modifies/cleaves the

Wnt ligand (Zhang et al., 2012). Tiki and Notum represent a

unique group of extracellular Wnt inhibitors with catalytic capac-

ities. We show that Notum is also aWnt-inactivating enzyme and

shares several commonalities with Tiki in modifying Wnt sub-

strates (Table 1): neither affectsWnt secretion; and either causes

Wnt to exhibit (1) faster electrophoreticmobility, (2) loss of hydro-

phobicity, (3) oxidized oligomer formation, and importantly, (4)

loss of receptor-binding and signaling activity (Figures 2, 3, S2,

and S4). Despite these similarities and that both Tiki and Notum

are hydrolytic enzymes, the natures of their Wnt-inactivating

modifications are different: Tiki cleaves the amino terminal resi-

dues of Wnt proteins as a protease (Zhang et al., 2012), whereas

Notum cleaves the palmitoleoylate adduct of Wnt proteins as a

deacylase (Figures 2 and 3). Therefore, distinct Wnt-inactivating

enzymes operate in modulating Wnt signaling, with Notum being

secreted and potentially diffusible, whereas Tiki being mem-

brane-tethered and affecting a cell autonomously or nearby

(Zhang et al., 2012). The recently determined structure of Notum

reveals an a/b hydrolase fold with a large hydrophobic cavity,

into which docking of a palmitoleoylate positions the acyl-oxy-

ester bond in proximity to the GxSxG catalytic center, providing

a structural basis for Notum deacylase function (Kakugawa et al.,

2015). Notum deacylates Wnt3a and possibly Wnt5a, but Notum

specificity towardWnt proteins and different Wnt signaling path-

ways remain to be studied.

Wnt Lipidation as a Requirement for Its Structure andActive Monomeric ConformationWnt palmitoleoylation has two critical functions; it may be

recognized by Wntless, a Wnt chaperone in the secretory

Inc.

Page 10: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

Figure 6. Notum Is Required for Neural In-

duction in Embryos and Animal Explants

(A) The Notum MOs reduced the expression

domain of Sox2, a pan-neural marker, and ex-

panded the expression domain of cytokeratin, an

epidermal marker, and the reciprocal changes

were rescued by mNotum, but not mNu-

tum(S239A), mRNA. See also Table S4.

(B) The Notum MOs suppressed expression of

neural markers induced by injection of the Chor-

din mRNA and restored cytokeratin expression

that were inhibited by Chordin. The effect of

Notum MOs was rescued by mNotum, but not

mNotum(S239A), mRNA; a cement gland marker,

XAG; anterior neural markers, Bf1 and Pax6;

a pan-neural marker, Sox2; a neuronal marker,

N-tubulin; an epidermal marker, Keratin; a

mesodermal marker, M-Actin; and a loading

control, ODC.

(C) Chordin induced Bf1 and Sox2 expression

when the Notum MOs and a b-catenin MO were

co-injected.

(D) Injection of Notum mRNA, like that of Chordin

mRNA, induced expression of neural markers

Sox2 and Pax6 and suppressed that of an epi-

dermal marker, cytokeratin, in animal cap ex-

plants.

See also Figure S6.

pathway (Coombs et al., 2010; Herr and Basler, 2012; Tang

et al., 2012), and it is essential for Wnt binding to Fz (Janda

et al., 2012). We show that Wnt depalmitoleoylation by

Notum occurs extracellularly, likely accounting for normal

secretion, but lack of receptor binding by deacylated Wnt3a

(Figures 1, 2, and 3). But unexpectedly, Wnt3a and Wnt5a de-

palmitoleoylated by Notum, similar to Wnt3a that is amino-

terminally cleaved by Tiki (Zhang et al., 2012), form large,

yet soluble, Wnt oligomers that are covalently linked by

inter-Wnt disulfide bonds (Figure 3). These oxidized Wnt

oligomers generated by Notum or Tiki appear to be similar

and are recapitulated by the Wnt3aDN mutant that lacks

Developmental Cell 32, 719–73

the amino terminal residues cleaved

by Tiki (Zhang et al., 2012). Therefore,

the Wnt oxidized oligomeric state (or

states), which is prevalent during Wnt

biogenesis (Zhang et al., 2012), may

be a default state of the Wnt protein.

Wnt palmitoleoylation thus appears to

have a third critical role; i.e., for main-

taining Wnt structural integrity to en-

sure its active monomeric conforma-

tion. The intact Wnt amino terminus,

which is rich in hydrophobic residues,

plays a similar role in maintaining the

Wnt fold (Zhang et al., 2012). The struc-

tural basis for the requirement of the

lipid, and of the Wnt amino terminus,

for the Wnt fold is not yet obvious

from the Wnt8 crystal structure (Janda

et al., 2012). Given that lipid modi-

fications of proteins are common in

signaling, a general role for lipid adducts in protein conforma-

tion deserves investigation.

Notum in Vertebrate Neural and Head Induction:Comparisons with the Role of TikiWhile Tiki1 is expressed in the Organizer and its descendent

head organizer during gastrulation (Zhang et al., 2012), Notum

(includingNotum’) ismaternally expressedand isenriched innaive

ectoderm (Figure 4).Overexpression ofNotumor Tiki1 dorsally re-

sults in head enlargement; depletion of Notum or Tiki1 causes

deficiency in head formation (Figure 5) (Zhang et al., 2012).

Thus, these two Wnt-inactivating enzymes are each required for

0, March 23, 2015 ª2015 Elsevier Inc. 727

Page 11: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

Table 2. Comparisons of Tiki1 and Notum Expression and

Functions in Xenopus Embryos

Tiki1 Notum

Expression

stages

from early

gastrulation

from egg throughout

embryogenesis

Early expression

patterns

organizer ectoderm, forming neural

plate, and anterior neural

plate border

Depletion

phenotypes

headless headless and neural plate

reduction

Action sites

(tissues)

head organizer ectoderm

Extracellular

features

membrane tethered

(non-diffusible)

secreted (diffusible?)

Table 1. Comparisons of Wnt3a Modifications by Tiki and Notum

Wnt3a Wnt3a + Tiki Wnt3a + Notum

Secretion normal normal normal

Hydrophobicity hydrophobic hydrophilic hydrophilic

Mobility in gel normal faster faster

Oxidized oligomer no yes yes

Signaling activity yes no no

Binding to receptor yes no no

Nature of

modification

� N-terminal

cleavage

deacylation

anterior development and not redundant. The action sites of

Notum and Tiki1 are distinct and complementary (Table 2). Tiki1

acts within the head organizer (Zhang et al., 2012), which induces

overlying ectoderm to form anterior neural tissues, whereas

Notum primarily acts within ectoderm to endow it with compe-

tence to become anterior brain, and Notum depletion does not

affect Organizer formation (Figures 5 and S5; Table 2).

The action of Notum within ectoderm appears to explain its

significant, but unique, role among Wnt antagonists in neural in-

duction. Upon Notum depletion, the expression of anterior and

pan neural markers is reduced, while that of an epidermal marker

is expanded (Figure 6), suggesting neural to epidermal fate con-

version. Naive ectoderm with Notum depletion loses its compe-

tence to respond to neural inducers/BMP antagonists (Figures 6

and S6). Conversely, overexpression of Notum, like that of Dkk1

(Fuentealba et al., 2007; Glinka et al., 1998), induces anterior

neural tissues (Figure 6). Thus, Wnt inactivation by Notum within

ectoderm is a critical part of the ‘‘default’’ neural fate (De Rob-

ertis and Kuroda, 2004; Ozair et al., 2013; Stern, 2005). This

requirement of Wnt inhibition by Notum, in addition to that of

BMP inhibition (and of FGF activation), for neuralization in Xeno-

pus seems to mirror neural induction mechanisms described in

the chick (Stern, 2005; Streit et al., 1998, 2000; Wilson et al.,

2000, 2001), supporting a unifying model for neural fate determi-

nation in vertebrates. The dependence onWnt and BMP co-inhi-

bition (and FGF activation) for neural induction may act through

parallel signaling pathways and/or through crosstalks by these

pathways (Fuentealba et al., 2007; Pera et al., 2003).

Notum exhibits dynamic expression in naive ectoderm in blas-

tula, in the forming neural plate in early gastrula, and at the ante-

rior border of (and exclusion from) the neural plate in late gastrula

(Figure 4). Interestingly, its ectodermal expression encompasses

the so-called blastula Chordin and Noggin expression (BCNE)

center, which represents the dorsal ectoderm giving rise to

future fore-, mid-, and hindbrain (Kuroda et al., 2004). The tran-

sient ectodermal Chordin and Noggin expression in blastula is

required for dorsal ectoderm to become anterior neural tissues

that are reinforced by Chordin and Noggin from the Organizer

during gastrulation (Kuroda et al., 2004). Similarly, we propose

that ectodermal Notum expression in blastula primes the dor-

sal ectoderm for sustained neuralization during gastrulation by

Organizer-generated BMP and Wnt antagonists, thereby under-

lying its requirement for neural and anterior induction. Subse-

quent Notum expression in the anterior neural plate border

may further reinforce Wnt inactivation for patterning forebrain

and surrounding regions such as pre-placodal ectoderm, which

728 Developmental Cell 32, 719–730, March 23, 2015 ª2015 Elsevier

is induced to form placodes through Wnt inhibition (Litsiou et al.,

2005). Experiments will be required to dissect the temporal

order and potentiallymultiple roles of Notumduring neural induc-

tion and patterning. Our results demonstrate that distinct Wnt

inactivation mechanisms by Notum in naive ectoderm and Tiki

in Organizer coordinate early brain formation, and that Notum

function in head development is conserved from planarians to

vertebrates. Notum as a Wnt inactivating enzyme represents a

potential therapeutic target.

EXPERIMENTAL PROCEDURES

Plasmids

Themouse Notum cDNA in pCAGGS contains the full coding region fusedwith

a33FLAG tagandwas fromDrs. S. Park andS. Sockanathan.GPC3andGPC4

expressing vectors were from A. Kikuchi. Notum coding region fused with a

13FLAG tag was subcloned into pCS2+. Notum(S239A) was generated by

PCR-based mutagenesis method based on pCS2+/Notum-FLAG. Notum-TM

andNotum(S239A)-TMwere constructedby inserting a cDNA fragment encod-

ing the transmembrane domain of epidermal growth factor receptor (EGFR)

(IATGMVGALLLLLVVALGIGLFM) between Notum and FLAG tag. Xenopus

laevis Notum and Notum’ cDNAs were amplified from embryo cDNAs via RT-

PCR and cloned into pCS2+. Details of plasmids are available upon request.

Triton X-114 Phase Separation

Triton X-114 phase separation was performed as described (Zhang et al.,

2012). Briefly, for CM separation, Wnt3a or Wnt5a CM was mixed with equal

volume of Triton X-114 buffer (10 millimolar (mM) Tris-HCl [pH 7.4], 150 mM

NaCl, and 4.5% Triton X-114); for whole cell lysate (WCL) separation, WCL

containing 1% Triton X-114 was mixed with equal volume of 3.5% Triton X-

114 buffer. The mixtures were incubated on ice for 5 min and then at 37�Cfor 5 min. After centrifugation at 2,000 g for 5 min, the top aqueous phase,

the bottom detergent phase, and the original mixture (total) were analyzed

by SDS-PAGE and western blotting.

Metabolic Labeling and Click-Chemistry

Metabolic labeling and click-chemistry assays were performed as described

(Zhang et al., 2012). Briefly, HEK293T cells or HEK293T cells stably expressing

hemagglutinin (HA)-Wnt3a (Zhang et al., 2012) in 60 millimeters (mm) tissue

culture dishes were transfected with indicated plasmids. At 24 hr after trans-

fection, cells were washed with serum-free DMEM once and then cultured in

the labeling medium (DMEM containing 5% dialyzed fetal bovine serum

[FBS], 40 mM az-15, or alk-14) for 12 hr. CM was collected and cleared by

centrifugation. HA-Wnt3a was enriched fromCMby anti-HA beads. The beads

were washed and re-suspended in 20 ml of PBS plus 2.25 ml freshly premixed

click-chemistry reaction mixture (alk-Rho or az-Rho, 100 mM, Tris(2-carbox-

yethyl)phosphine [TCEP], 1 mM, Tris[(1-benzyl-1H-1,2,3-triazol-4-yl)methyl]

Inc.

Page 12: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

amine [TBTA], 100 mM, and CuSO4$5H2O, 1mM) for 1 hr at room temperature.

The samples were denatured and separated by SDS-PAGE, and the gels were

fixed and scanned on a GE Healthcare Typhoon 9400 variable-mode imager

for Rhodamine-associated signal at excitation 532 nanometers (nm)/emission

580 nm. The same samples were also subjected to SDS-PAGE and western

blotting to detect total Wnt3a protein.

Protein Purification and In Vitro Deacylation of Wnt3a

The CM from HEK293T cells expressing Notum-FLAG or Notum(S239A)-

FLAG was mixed with anti-FLAG beads and incubated at 4�C overnight.

The beads were washed with PBS/0.1% Triton X-100 and the FLAG fusion

proteins were eluted with elution buffer (50 mM HEPES (pH 7.4), 100 mM

NaCl, 0.1% Triton X-100, and 50 mg/ml 33FLAG peptide). For in vitro deacy-

lation, alk-14 labeled Wnt3a in CM was immunoprecipitated with anti-HA

agarose beads and incubated with purified Notum or Notum(S239A) protein

at room temperature overnight, followed with click-chemistry reaction. The

reaction mixtures were denatured and separated by SDS-PAGE. The fluores-

cent signal and total Wnt3a proteins were detected by in-gel scanning and

western blotting.

Xenopus Embryo Manipulations

Frogs (Xenopus laevis) were maintained at the Animal Resource Children’s

Hospital (ARCH) in accordance with institutional guidelines. Procedures for

embryo manipulation, RT-PCR, and in situ hybridization were performed as

previously described (Zhang et al., 2012). The full-length Xenopus laevis

Notum’ cDNA was used to make in situ probe.

mRNA and Morpholino Injection

Axis duplication and animal cap assays were performed as described (Zhang

et al., 2012). For axis duplication assays, Xwnt8 (1 picogram [pg]) or b-catenin

(50 pg) mRNA was injected alone or together with Notum (200 pg) mRNA into

the ventral marginal zone at the 4- or 8-cell stage, and the phenotype was

scored at the tadpole stage. GFP was used as a negative control. For animal

cap assays, indicatedmRNAs (Notum, 100 and 200 pg; Xwnt8, 5 pg; b-catenin

30 pg; Xnr1, 250 pg; BMP4, 100 pg; and Chordin, 200 pg) and morpholinos (20

nanograms [ng]) were injected into the animal pole at the 4-cell stage, and an-

imal caps were dissected at stage 9 (in the case of Xwnt8, Xnr1, BMP4, and

Chordin), or at stage 8.5 and treated with recombinant proteins (bFGF,

100 ng/ml and Noggin, 500 ng/ml) until stage 11 (for Xwnt8, Xnr1, BMP4,

and bFGF) or stage 18 (for Noggin or Chordin) before RT-PCR. To examine

Notum and Notum’ MO specificity, 500 pg of xNotum, xNotum’, or mNotum

mRNAwas injected with control MO,Notum, orNotum’MO (20 ng) into the an-

imal pole at the 2-cell stage and cultured until stage 10 for western blotting. To

knockdown the endogenous xNotum and xNotum’, 20 ng of control MO or

Notum and Notum’ MOs were injected into two dorso-animal blastomeres at

the 8-cell stage, or two dorsal A1 or B1 cells at the 32-cell stage, and the

phenotype was scored at stage 35. To rescue the MOs (xNotum and xNotum’)

phenotype, 200 pg of mNotum, 20 pg of DKK1 mRNA, or 10 ng of b-catenin

MO was injected together with MOs.

ACCESSION NUMBERS

The GenBank accession numbers for the Xenopus laevis Notum, Notum’, and

Notum2 reported in this paper are KP781855, KP781856, and KP781857,

respectively.

SUPPLEMENTAL INFORMATION

Supplemental Information includes Supplemental Experimental Procedures,

six figures, and four tables and can be found with this article online at http://

dx.doi.org/10.1016/j.devcel.2015.02.014.

AUTHOR CONTRIBUTIONS

X.Z. performed all biochemical experiments. S.-M.C. performed most Xeno-

pus experiments. N.G.A., A.H.R., and J.G.A. performed additional Xenopus

experiments. B.T.M. contributed to experiments in mammalian cells and per-

Develo

formed bioinformatic analysis. X.Z., S.-M.C., J.G.A., M.Z., E.Y.J., and X.H.

contributed to experimental designs, X.Z., S.-M.C., and X.H. wrote the manu-

script, and all co-authors edited the manuscript.

ACKNOWLEDGMENTS

We thank S. Park, S. Sockanathan, and A. Kikuchi for plasmids, J.-P. Saint-

Jeannet for comments, and J.-P. Vincent for discussion. X.Z. and S.-M.C.

were in part supported by a grant from Harvard William Randolph Hearst

Fund and a fellowship from National Research Foundation of Korea, respec-

tively. N.G.A. is in part supported by a postdoctoral fellowship from Science

Without Borders program/Council for Research and Technology (CNPq) of

Brazil. M.Z. held a Marie Curie Intra European Fellowship (IEF) fellowship.

J.G.A. and A.H.R. are supported by CNPq and Rio de Janeiro State Founda-

tion for Science support (FAPERJ) of Brazil. E.Y.J. acknowledges support by

Cancer Research UK (A10976) and by the Wellcome Trust Centre for Human

Genetics (090532/Z/09/Z). X.H. acknowledges support by NIH (RO1-

GM057603) and by Boston Children’s Hospital Intellectual and Developmental

Disabilities Research Center (P30 HD-18655). X.H. is an American Cancer So-

ciety Research Professor.

Received: January 8, 2015

Revised: February 17, 2015

Accepted: February 17, 2015

Published: March 12, 2015

REFERENCES

Alexandre, C., Baena-Lopez, A., and Vincent, J.P. (2014). Patterning and

growth control by membrane-tethered Wingless. Nature 505, 180–185.

Barrott, J.J., Cash, G.M., Smith, A.P., Barrow, J.R., andMurtaugh, L.C. (2011).

Deletion of mouse Porcn blocks Wnt ligand secretion and reveals an ecto-

dermal etiology of human focal dermal hypoplasia/Goltz syndrome. Proc.

Natl. Acad. Sci. USA 108, 12752–12757.

Beckett, K., Franch-Marro, X., and Vincent, J.P. (2008). Glypican-mediated

endocytosis of Hedgehog has opposite effects in flies and mice. Trends Cell

Biol. 18, 360–363.

Biechele, S., Cox, B.J., and Rossant, J. (2011). Porcupine homolog is required

for canonical Wnt signaling and gastrulation in mouse embryos. Dev. Biol. 355,

275–285.

Cantu, J.A., Flowers, G.P., and Topczewski, J. (2013). Notum homolog plays a

novel role in primary motor innervation. J. Neurosci. 33, 2177–2187.

Capurro, M., Martin, T., Shi, W., and Filmus, J. (2014). Glypican-3 binds to

Frizzled and plays a direct role in the stimulation of canonical Wnt signaling.

J. Cell Sci. 127, 1565–1575.

Clevers, H., and Nusse, R. (2012). Wnt/b-catenin signaling and disease. Cell

149, 1192–1205.

Coombs, G.S., Yu, J., Canning, C.A., Veltri, C.A., Covey, T.M., Cheong, J.K.,

Utomo, V., Banerjee, N., Zhang, Z.H., Jadulco, R.C., et al. (2010). WLS-depen-

dent secretion of WNT3A requires Ser209 acylation and vacuolar acidification.

J. Cell Sci. 123, 3357–3367.

Cormier, R.T., Hong, K.H., Halberg, R.B., Hawkins, T.L., Richardson, P.,

Mulherkar, R., Dove, W.F., and Lander, E.S. (1997). Secretory phospholipase

Pla2g2a confers resistance to intestinal tumorigenesis. Nat. Genet. 17,

88–91.

Cruciat, C.M., and Niehrs, C. (2013). Secreted and transmembrane wnt inhib-

itors and activators. Cold Spring Harb. Perspect. Biol. 5, a015081.

Dale, L., and Slack, J.M. (1987). Fate map for the 32-cell stage of Xenopus lae-

vis. Development 99, 527–551.

De Robertis, E.M., and Kuroda, H. (2004). Dorsal-ventral patterning and neural

induction in Xenopus embryos. Annu. Rev. Cell Dev. Biol. 20, 285–308.

Delaune, E., Lemaire, P., and Kodjabachian, L. (2005). Neural induction in

Xenopus requires early FGF signalling in addition to BMP inhibition.

Development 132, 299–310.

Filmus, J., Capurro, M., and Rast, J. (2008). Glypicans. Genome Biol. 9, 224.

pmental Cell 32, 719–730, March 23, 2015 ª2015 Elsevier Inc. 729

Page 13: Notum Is Required for Neural and Head Induction via Wnt … · 2017-02-24 · Developmental Cell Article Notum Is Required for Neural and Head Induction via Wnt Deacylation, Oxidation,

Flowers, G.P., Topczewska, J.M., and Topczewski, J. (2012). A zebrafish

Notum homolog specifically blocks the Wnt/b-catenin signaling pathway.

Development 139, 2416–2425.

Fuentealba, L.C., Eivers, E., Ikeda, A., Hurtado, C., Kuroda, H., Pera, E.M., and

De Robertis, E.M. (2007). Integrating patterning signals: Wnt/GSK3 regulates

the duration of the BMP/Smad1 signal. Cell 131, 980–993.

Gerlitz, O., and Basler, K. (2002). Wingful, an extracellular feedback inhibitor of

Wingless. Genes Dev. 16, 1055–1059.

Giraldez, A.J., Copley, R.R., and Cohen, S.M. (2002). HSPG modification by

the secreted enzyme Notum shapes the Wingless morphogen gradient. Dev.

Cell 2, 667–676.

Glinka, A., Wu, W., Delius, H., Monaghan, A.P., Blumenstock, C., and Niehrs,

C. (1998). Dickkopf-1 is a member of a new family of secreted proteins and

functions in head induction. Nature 391, 357–362.

Han, C., Yan, D., Belenkaya, T.Y., and Lin, X. (2005). Drosophila glypicans

Dally and Dally-like shape the extracellular Wingless morphogen gradient in

the wing disc. Development 132, 667–679.

Hannoush, R.N. (2012). Profiling cellular myristoylation and palmitoylation us-

ing u-alkynyl fatty acids. Methods Mol. Biol. 800, 85–94.

Harland, R., and Gerhart, J. (1997). Formation and function of Spemann’s

organizer. Annu. Rev. Cell Dev. Biol. 13, 611–667.

Hausmann, G., Banziger, C., and Basler, K. (2007). Helping Wingless take

flight: how WNT proteins are secreted. Nat. Rev. Mol. Cell Biol. 8, 331–336.

Heasman, J., Kofron, M., and Wylie, C. (2000). Beta-catenin signaling activity

dissected in the early Xenopus embryo: a novel antisense approach. Dev. Biol.

222, 124–134.

Heeg-Truesdell, E., and LaBonne, C. (2006). Neural induction in Xenopus re-

quires inhibition of Wnt-beta-catenin signaling. Dev. Biol. 298, 71–86.

Herr, P., and Basler, K. (2012). Porcupine-mediated lipidation is required for

Wnt recognition by Wls. Dev. Biol. 361, 392–402.

Hikasa, H., and Sokol, S.Y. (2013). Wnt signaling in vertebrate axis specifica-

tion. Cold Spring Harb. Perspect. Biol. 5, a007955.

Janda, C.Y., Waghray, D., Levin, A.M., Thomas, C., and Garcia, K.C. (2012).

Structural basis of Wnt recognition by Frizzled. Science 337, 59–64.

Kakugawa, S., Langton, P.F., Zebisch, M., Howell, S.A., Chang, T.H., Liu, Y.,

Feizi, T., Bineva, G., O’Reilly, N., Snijders, A.P., et al. (2015). Notum deacylates

Wnt proteins to suppress signalling activity. Nature, in press. Published online

February 25, 2015. http://dx.doi.org/10.1038/nature14259.

Kengaku, M., and Okamoto, H. (1995). bFGF as a possible morphogen for the

anteroposterior axis of the central nervous system in Xenopus. Development

121, 3121–3130.

Kiecker, C., and Niehrs, C. (2001). A morphogen gradient of Wnt/beta-catenin

signalling regulates anteroposterior neural patterning in Xenopus.

Development 128, 4189–4201.

Komekado, H., Yamamoto, H., Chiba, T., and Kikuchi, A. (2007). Glycosylation

and palmitoylation of Wnt-3a are coupled to produce an active form ofWnt-3a.

Genes Cells 12, 521–534.

Kreuger, J., Perez, L., Giraldez, A.J., and Cohen, S.M. (2004). Opposing activ-

ities of Dally-like glypican at high and low levels of Wingless morphogen activ-

ity. Dev. Cell 7, 503–512.

Kuroda, H., Wessely, O., and De Robertis, E.M. (2004). Neural induction in

Xenopus: requirement for ectodermal and endomesodermal signals via

Chordin, Noggin, beta-Catenin, and Cerberus. PLoS Biol. 2, E92.

Lamb, T.M., and Harland, R.M. (1995). Fibroblast growth factor is a direct neu-

ral inducer, which combined with noggin generates anterior-posterior neural

pattern. Development 121, 3627–3636.

Litsiou, A., Hanson, S., and Streit, A. (2005). A balance of FGF, BMP and WNT

signalling positions the future placode territory in the head. Development 132,

4051–4062.

MacDonald, B.T., and He, X. (2012). Frizzled and LRP5/6 receptors for Wnt/

b-catenin signaling. Cold Spring Harb. Perspect. Biol. 4, 4.

MacDonald, B.T., Tamai, K., and He, X. (2009). Wnt/beta-catenin signaling:

components, mechanisms, and diseases. Dev. Cell 17, 9–26.

730 Developmental Cell 32, 719–730, March 23, 2015 ª2015 Elsevier

Marchal, L., Luxardi, G., Thome, V., and Kodjabachian, L. (2009). BMP inhibi-

tion initiates neural induction via FGF signaling and Zic genes. Proc. Natl.

Acad. Sci. USA 106, 17437–17442.

Moody, S.A. (1987). Fates of the blastomeres of the 32-cell-stage Xenopus

embryo. Dev. Biol. 122, 300–319.

Nardini, M., and Dijkstra, B.W. (1999). Alpha/beta hydrolase fold enzymes: the

family keeps growing. Curr. Opin. Struct. Biol. 9, 732–737.

Ozair, M.Z., Kintner, C., and Brivanlou, A.H. (2013). Neural induction and early

patterning in vertebrates. Wiley interdisciplinary reviews. Dev. Biol. 2,

479–498.

Pera, E.M., Ikeda, A., Eivers, E., and De Robertis, E.M. (2003). Integration of

IGF, FGF, and anti-BMP signals via Smad1 phosphorylation in neural induc-

tion. Genes Dev. 17, 3023–3028.

Petersen, C.P., and Reddien, P.W. (2011). Polarized notum activation at

wounds inhibits Wnt function to promote planarian head regeneration.

Science 332, 852–855.

Proffitt, K.D., and Virshup, D.M. (2012). Precise regulation of porcupine activity

is required for physiological Wnt signaling. J. Biol. Chem. 287, 34167–34178.

Rios-Esteves, J., and Resh, M.D. (2013). Stearoyl CoA desaturase is required

to produce active, lipid-modified Wnt proteins. Cell Rep. 4, 1072–1081.

Rios-Esteves, J., Haugen, B., and Resh, M.D. (2014). Identification of key res-

idues and regions important for porcupine-mediated Wnt acylation. J. Biol.

Chem. 289, 17009–17019.

Sakane, H., Yamamoto, H., Matsumoto, S., Sato, A., and Kikuchi, A. (2012).

Localization of glypican-4 in different membrane microdomains is involved in

the regulation of Wnt signaling. J. Cell Sci. 125, 449–460.

Stern, C.D. (2005). Neural induction: old problem, new findings, yetmore ques-

tions. Development 132, 2007–2021.

Streit, A., Lee, K.J., Woo, I., Roberts, C., Jessell, T.M., and Stern, C.D. (1998).

Chordin regulates primitive streak development and the stability of induced

neural cells, but is not sufficient for neural induction in the chick embryo.

Development 125, 507–519.

Streit, A., Berliner, A.J., Papanayotou, C., Sirulnik, A., and Stern, C.D. (2000).

Initiation of neural induction by FGF signalling before gastrulation. Nature 406,

74–78.

Strigini, M., and Cohen, S.M. (2000). Wingless gradient formation in the

Drosophila wing. Curr. Biol. 10, 293–300.

Takada, R., Satomi, Y., Kurata, T., Ueno, N., Norioka, S., Kondoh, H., Takao,

T., and Takada, S. (2006). Monounsaturated fatty acidmodification ofWnt pro-

tein: its role in Wnt secretion. Dev. Cell 11, 791–801.

Tang, X., Wu, Y., Belenkaya, T.Y., Huang, Q., Ray, L., Qu, J., and Lin, X. (2012).

Roles of N-glycosylation and lipidation in Wg secretion and signaling. Dev.

Biol. 364, 32–41.

Willert, K., Brown, J.D., Danenberg, E., Duncan, A.W., Weissman, I.L., Reya,

T., Yates, J.R., 3rd, and Nusse, R. (2003). Wnt proteins are lipid-modified

and can act as stem cell growth factors. Nature 423, 448–452.

Wilson, S.I., Graziano, E., Harland, R., Jessell, T.M., and Edlund, T. (2000). An

early requirement for FGF signalling in the acquisition of neural cell fate in the

chick embryo. Curr. Biol. 10, 421–429.

Wilson, S.I., Rydstrom, A., Trimborn, T., Willert, K., Nusse, R., Jessell, T.M.,

and Edlund, T. (2001). The status of Wnt signalling regulates neural and

epidermal fates in the chick embryo. Nature 411, 325–330.

Yan, D., and Lin, X. (2009). Shaping morphogen gradients by proteoglycans.

Cold Spring Harb. Perspect. Biol. 1, a002493.

Zecca, M., Basler, K., and Struhl, G. (1996). Direct and long-range action of a

wingless morphogen gradient. Cell 87, 833–844.

Zeidman, R., Jackson, C.S., and Magee, A.I. (2009). Protein acyl thioesterases

(Review). Mol. Membr. Biol. 26, 32–41.

Zhang, X., Abreu, J.G., Yokota, C., MacDonald, B.T., Singh, S., Coburn, K.L.,

Cheong, S.M., Zhang, M.M., Ye, Q.Z., Hang, H.C., et al. (2012). Tiki1 is

required for head formation via Wnt cleavage-oxidation and inactivation. Cell

149, 1565–1577.

Inc.